Edinburgh Research Explorer

Size: px
Start display at page:

Download "Edinburgh Research Explorer"

Transcription

1 Edinburgh Research Explorer Characterization of a novel simian immunodeficiency virus (SIV) from L'Hoest monkeys (Cercopithecus l'hoesti) Citation for published version: Hirsch, VM, Campbell, BJ, Bailes, E, Goeken, R, Brown, C, Elkins, WR, Axthelm, M, Murphey-Corb, M & Sharp, PM 1999, 'Characterization of a novel simian immunodeficiency virus (SIV) from L'Hoest monkeys (Cercopithecus l'hoesti): Implications for the origins of SIVmnd and other primate lentiviruses' Journal of Virology, vol 73, no. 2, pp Link: Link to publication record in Edinburgh Research Explorer Document Version: Publisher's PDF, also known as Version of record Published In: Journal of Virology Publisher Rights Statement: Free in PMC. General rights Copyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorer content complies with UK legislation. If you believe that the public display of this file breaches copyright please contact openaccess@ed.ac.uk providing details, and we will remove access to the work immediately and investigate your claim. Download date: 28. Apr. 2017

2 JOURNAL OF VIROLOGY, Feb. 1999, p Vol. 73, No X/99/$ Copyright 1999, American Society for Microbiology. All Rights Reserved. Characterization of a Novel Simian Immunodeficiency Virus (SIV) from L Hoest Monkeys (Cercopithecus l hoesti): Implications for the Origins of SIVmnd and Other Primate Lentiviruses VANESSA M. HIRSCH, 1 * BARBARA J. CAMPBELL, 1 ELIZABETH BAILES, 2 ROBERT GOEKEN, 1 CHARLES BROWN, 1 WILLIAM R. ELKINS, 3 MICHAEL AXTHELM, 4 MICHAEL MURPHEY-CORB, 5 AND PAUL M. SHARP 2 Laboratory of Molecular Microbiology 1 and Laboratory of Infectious Diseases, 3 National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852; Division of Genetics, University of Nottingham, Queens Medical Center, Nottingham NG7 2UH, United Kingdom 2 ; Division of Pathobiology, Oregon Regional Primate Research Center, Beaverton, Oregon ; and Department of Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania 5 Received 13 August 1998/Accepted 20 October 1998 The human immunodeficiency virus types 1 and 2 (HIV-1 and HIV-2) appear to have originated by crossspecies transmission of simian immunodeficiency virus (SIV) from asymptomatically infected African primates. Few of the SIVs characterized to date efficiently infect human primary lymphocytes. Interesting, two of the three identified to infect such cultures (SIVsm and SIVcpz) have appeared in human populations as genetically related HIVs. In the present study, we characterized a novel SIV isolate from an East African monkey of the Cercopithecus genus, the l hoest monkey (C. l hoesti), which we designated SIVlhoest. This SIV isolate efficiently infected both human and macaque lymphocytes and resulted in a persistent infection of macaques, characterized by high primary virus load and a progressive decline in circulating CD4 lymphocytes, consistent with progression to AIDS. Phylogenetic analyses showed that SIVlhoest is genetically distinct from other previously characterized primate lentiviruses but clusters in the same major lineage as SIV from mandrills (SIVmnd), a West African primate species. Given the geographic distance between the ranges of l hoest monkeys and mandrills, this may indicate that SIVmnd arose through cross-species transmission from close relatives of l hoest monkeys that are sympatric with mandrills. These observations lend support to the hypothesis that the primate lentiviruses originated and coevolved within monkeys of the Cercopithecus genus. Regarded in this light, lentivirus infections of primates not belonging to the Cercopithecus genus may have resulted from cross-species transmission in the not-too-distant past. The human immunodeficiency virus types 1 and 2 (HIV-1 and HIV-2) are lentiviruses that appear to have originated by cross-species transmission from African primates (22, 25, 45). Five distinct types of lentiviruses from nonhuman primates have previously been molecularly characterized: SIVsm from sooty mangabeys (Cercocebus atys) (7, 19, 37, 44), SIVagm from the four species within the African green monkey superspecies (Cercopithecus aethiops) (3, 4, 8, 10, 11, 20, 28, 30, 38), SIVsyk from Sykes monkeys (Cercopithecus mitis) (9, 21), SIVmnd from mandrills (Mandrillus sphinx) (49), and SIVcpz from chimpanzees (Pan troglodytes) (26, 27, 43, 50). It is currently believed that these characterized viruses may represent just a small part of a very large family of primate lentiviruses. Indeed, serologic surveys of other African primates have identified a number of other monkey species that have SIV-specific antibodies (18, 33, 36, 40, 42). For example, SIV has recently been isolated from a red-capped mangabey (Cercocebus torquatus torquatus); partial characterization of this virus suggests that it may represent a distinct (sixth) lineage, although analysis of the complete genome will be necessary to establish the * Corresponding author. Mailing address: Laboratory of Molecular Microbiology, NIAID, NIH, Parklawn Dr., Rockville, MD Phone: (301) Fax: (301) exact phylogenetic relationship between SIVrcm and other primate lentiviruses (17). Further study of the lentiviruses infecting nonhuman primates is important because it may provide insight into the origins and evolution of HIV in humans. The phylogenetic relationships among SIVsm and HIV-2 isolates clearly implicate SIVsm as the proximal source of the HIV-2 epidemic in West Africa (13, 14, 19, 37), but the origins of HIV-1 have not been identified with certainty. A small number of pet chimpanzees have been found to be infected with a virus (SIVcpz) closely related to HIV-1 (26, 27, 43, 50), but the lack of serologic evidence of SIVcpz infection in feral chimpanzee populations sheds doubt on whether this virus constitutes a natural infection in this species (45). It is intriguing that SIVs which are capable of infecting human peripheral blood mononuclear cells (PBMC) are in a minority and at least for two of these (SIVsm and SIVcpz), related HIVs (HIV-2 and HIV-1, respectively) have been demonstrated in humans. The ability to infect the CD4 lymphocytes of humans may thus be a prerequisite for cross-species transmission to humans. While the ability of these SIV strains to utilize human coreceptors is clearly one mechanism, the accessory proteins Vpr and Vif also limit the ability of some SIVs to replicate in human PBMC (46, 47). Among the other species of African primates identified by serologic surveys as harboring SIV are a number of species 1036

3 VOL. 73, 1999 NOVEL SIV FROM L HOEST MONKEYS 1037 from the genus Cercopithecus, commonly called the guenons (32, 34, 41). Cercopithecus monkeys are a diverse group of 25 species and as many as 70 subspecies of forest-dwelling monkeys that are distributed throughout subsaharan Africa (32, 34, 41). The most commonly known members are the four species of African green monkeys (sabaeus monkeys, C. sabaeus; grivets, C. aethiops; vervets, C. pygerythrus; and tantalus monkeys, C. tantalus). Each of these four species harbors closely related SIV strains, leading to the hypothesis that the primate lentiviruses have coevolved with their host species (22, 25, 45). The only other member of the guenons from which a novel SIV has been characterized is the Sykes monkey from which SIVsyk (9, 21) was isolated. Despite the fact that both Sykes and African green monkeys are members of the same genus, SIVsyk and SIVagm are no more closely related to one another than to any of the other characterized primate lentiviruses. Other Cercopithecus monkeys for which SIV seropositivity has been observed include DeBrazza monkeys (C. neglectus) (40), redtailed monkeys (C. ascanius schmidtii) (20), Hamlyn s monkeys (C. hamlyni) (40), and l hoest monkeys (C. l hoesti l hoesti; 40). The close phylogenetic relationships among these monkeys and their widespread distribution across Africa, often in distinct habitats, suggest that the study of SIV in these species will enrich our understanding of the evolution of the primate lentiviruses in general and of the origins of the AIDS epidemic in humans in particular. Molecular characterization of SIV strains from many of the African monkeys has been hampered by the lack of availability of samples from feral or wild-caught animals and the difficulty in isolating the virus. An alternative source of samples are wild-caught captive populations, such as those found at primate centers or zoo collections, although it is difficult to extrapolate that seropositivity in these populations is indicative of similar infection in free-living feral populations. In a serologic survey of a troop of l hoest monkeys (C. l hoesti l hoesti) in the Portland Zoo, one wild-caught male with antibodies cross-reactive with SIVmac was identified. Here we describe the isolation of SIV from PBMC of this seropositive l hoest monkey by cocultivation with the human T-cell line, Molt4 Clone 8 (M4C8). This isolate, designated SIVlhoest, has been characterized and compared to other known SIV sequences. MATERIALS AND METHODS Virus isolation and infectivity studies. Virus was isolated from PBMC of a male l hoest monkey by coculture of phytohemagglutinin-stimulated PBMC with M4C8 cells, using production of reverse transcriptase (RT) activity in the culture supernatant as a measure of viral replication. Virus stocks were prepared from these infected cells by filtration through a m-pore-size filter and cryopreserved in the vapor phase of liquid nitrogen for use in subsequent infectivity studies. These culture supernatants were used to infect M4C8 and CEMss cells to establish a cell line for isolation of total genomic DNA for subsequent cloning studies and for preparation of cell-free virus stocks for infectivity studies in vitro and in macaques. Animal infectivity studies. Four juvenile (simian retrovirus- and simian T-cell leukemia virus type 1-free) pigtailed macaques (Macaca nemestrina) were inoculated intravenously with 1 ml of the uncloned SIVlhoest virus stock described above. Macaques were handled in accordance with the guidelines of the NIH Animal Care and Use Committee. Animals were subsequently monitored by virus isolation, coculture of PBMC (at 1, 2, 3, 4, 8, 12, and 16 weeks), limiting dilution infectivity assay of plasma (at 1, 2, 3, and 4 weeks), limiting dilution coculture of disrupted lymph node cells with CEMss cells (at 1, 2, 4, and 16 weeks), and in situ hybridization (ISH) of lymph node biopsies for SIVlhoest viral RNA expression. Antibody responses were monitored by Western blot analysis, using SIVlhoest virus pelleted through sucrose as the viral antigen and published Western blot procedures (24). Lymphocyte subsets (CD4, CD8, CD2, and CD20) were analyzed at the same intervals as virus isolation assays from PBMC, and hematological alterations were monitored by performing complete blood counts. PCR amplification and plasmid cloning. Total cellular DNA was extracted from infected CEMss cells at 10 to 15 days postinfection. Degenerate primers (LV1 and LV2 for the first round and LV3 and DDMY for the second round of amplification) designed to PCR amplify a small portion of all lentivirus pol sequences (15) were used to amplify a portion of pol from total cellular DNA. The amplification conditions were as follows: one cycle at 94 C for 2 min, 37 C for 2 min, and 72 C for 3 min, followed by 35 cycles at 94 C for 30 s, 55 C for 30 s, and 72 C for 1 min. The resulting 120-bp fragment was cloned into the TA plasmid vector (Invitrogen) and sequenced. A reverse primer was designed based on this sequence and by using a conserved forward primer situated in the primer binding site of SIVs (30), a 2.4-kb fragment was amplified using the following PCR amplification conditions: 94 C for 30 s, 55 C for 30 s, and 72 C for 2 min. This fragment was cloned into the plasmid vector pgem-7zf and subsequently utilized as a probe for Southern blot hybridization and screening of a bacteriophage lambda library. Bacteriophage lambda cloning. A variety of restriction enzymes were evaluated by Southern blot hybridization of total DNA from infected CEMss cells using the 2.4-kb gag-pol virus fragment described above as a probe. Based on this analysis, SstI was chosen for full-length cloning since the proviral DNA did not appear to contain any SstI sites. Total cellular DNA was digested to completion with SstI, fractionated over a 20 to 60% sucrose gradient to obtain 9- to 20-kb fragments, and ligated into SstI- and XhoI-cleaved arms of Gem12 (Promega, Madison, Wis.). Ligation products were packaged in vitro (Gigapack Gold III; Stratagene), titrated, and plated on bacteria (Escherichia coli K802). A total of recombinant plaques were screened, using a horseradish peroxidaselabeled 2.4-kb gag-pol fragment and a direct detection method and following the manufacturer s instructions (ECL Direct Detection; Amersham). One positive clone ( SIVlhoest-P7) was detected and plaque purified. To facilitate sequence analysis, three subgenomic clones were generated by digestion of the recombinant lambda clone with SstI and BamHI and ligation into a pgem-7zf vector. The complete SIVlhoest provirus, including flanking cellular sequences (a 14-kb SstI fragment), was also subcloned into pgem-7zf to facilitate subsequent transfection studies. Both strands of the virus were sequenced by a combination of manual dideoxy sequencing using T4 sequenase (USB) and automated fluorescent sequencing (Taq amplification/termination; Perkin Elmer Applied Biosystems) in an ABI 377. Nucleotide sequence analysis was performed by using the GeneWorks (Oxford Molecular) and the Intelligenetics programs (Oxford Molecular). Sequence comparisons. The predicted protein sequences encoded by SIV lhoest were compared to the following representatives of the major primate lentivirus lineages: HIV-1 subtype A (isolate U455; GenBank accession no. M62320), subtype B (BRU; K02013), and group O (ANT70; L20587); SIVcpz strains Gab (X52154) and Ant (U42720); SIVsm (PBj; M31325); HIV-2 subtype A (ROD; M15390) and subtype B (UC1; L07625); SIVagm from vervets (ver155; M29975), grivets (gri-1; M58410), and tantalus monkeys (tan-1; U58991); SIVsyk (173; L06042); and SIVmnd (GB1; M27470). Protein sequences were aligned using CLUSTAL X (48) with minor subsequent adjustments using SEAVIEW (12). Sites that could not be aligned unambiguously, as well as all sites for which there was a gap in any of the sequences, were excluded from the analyses. The extent of sequence difference, along the genome, between SIVlhoest and other viruses was examined in a diversity plot in which protein sequences were concatenated with segments encoded by overlapping genomic regions represented only once: for example, in the region of the Gag-Pol overlap, the amino terminus of the Pol protein was excluded. The fractional amino acid sequence difference was calculated for a window size of 200 residues, moved in steps of 10 residues. The phylogenetic relationship of SIVlhoest to other primate lentivirus sequences was estimated from aligned Gag, Pol, and Env sequences and from subregions within these alignments to check for evidence that SIVlhoest might have a mosaic genome resulting from recombination during its ancestry. In the absence of such evidence, a summary phylogeny was derived from a concatenated Gag-Pol-Env alignment (the amino terminus of the Pol protein was again excluded) totaling 1,909 amino acids. Relationships were estimated by the neighbor-joining and maximum likelihood methods. The neighbor-joining method, with Kimura protein distances and 1,000 bootstrap replicates, was implemented within the CLUSTAL X package (48). The maximum likelihood method was implemented with PROTML (1) using the JTT model. The order of sequence input was shuffled five times, with the same best tree being found each time. Generation of ISH probes. PCR was used to amplify five 1.5- to 2-kb fragments of the SIVlhoest provirus from the complete plasmid clone by using the following primers, where the restriction sites (SstI and Csp45I) introduced to facilitate cloning are underlined: 1F, (nucleotide [nt] 1018) 5 -ttagagctcttgtgagaagtgtgtaa ttctgat; 1R, (nt 2482) 5 -tgattcgaatctgcttttgttggagcactctcc; 2F, (nt 2506) 5 -atcga gctcgagagcactggagacttacaggac; 2R, (nt 4505) 5 -atcttcgaattcctttatgagcaggcaccca tc; 3F, (nt 4529) 5 -attgagctctaggaggtaatcaagaggtagacc; 3R, (nt 6487) 5 -catttcg aataattaacactgcatacttaacat; 4F, (nt 6502) 5 -ttagagctcccaatactagcctctgggtcacga; 4R, (nt 8503) 5 -atattcgaataattaacactgcatacttaacat; 5F, (nt 8527) 5 -atcgagctctac ttgtaatcatagggttaagag; 5R, (nt 9844) 5 -taattcgaaactagcttgtactttctaacaatg. These five fragments were cloned into pgem-7zf, the subsequent clones were digested with SstI (antisense) and Csp45I (sense), and RNA was transcribed to incorporate digoxigenin using Sp6 and T7 polymerase, respectively. The pooled antisense probe was used to detect viral mrna expression in lymph node biopsy samples that were fixed in STRECKS fixative using previously described methodology (24).

4 1038 HIRSCH ET AL. J. VIROL. TABLE 1. Sequential virus isolation from PBMC and lymph nodes of macaques inoculated with SIVlhoest isolates Isolation of virus at indicated wk postinoculation a Isolate source Macaque PBMC Spleen a Virus isolation from PBMC and lymph nodes as detected by RT activity in culture supernatant within a 6-week period of coculture is indicated by a, and inability to isolate virus is indicated by a. FIG. 1. In vitro growth characteristics of SIVlhoest and other primate lentiviruses in human and macaque PBMC. The replication of SIVlhoest, SIVagm, SIVsm, SIVsyk, and HIV-1 as assessed by RT activity in culture supernatant is shown graphically for macaque (A) and human (B) PBMC. For this assay, equivalent amounts (based on RT activity of stocks) of the following viruses were used: SIVlhoest-P, SIVagm155-4, SIVsmE543-3, SIVsyk/cm173, and HIV-1 IIIB. Since RT activity was never observed in SIVsyk-infected cultures, the symbols are superimposed by values for the negative control. Transfection and infectivity studies. Virus stocks were generated by transfection of 5 to 10 g of either the lambda or plasmid clone into 293 cells by a calcium phosphate-mediated procedure (CellPhect; Stratagene). The infectivity of these filtered supernatants was evaluated by infection of CEMss cells, macaque PBMC, and macaque monocyte-derived macrophages as previously described (24). Nucleotide sequence accession number. The complete sequence of SIVlhoest has been submitted to GenBank under accession no. AF RESULTS Infectivity and pathogenicity of SIVlhoest in vivo. SIVlhoest was clearly distinct from other primate lentiviruses, such as SIVagm, SIVsm, or HIV-1, since the Gag antigens in culture supernatants of persistently infected cells did not cross-react with a commercially available SIVmac p27 antigen capture assay (Coulter Corp.). As a prelude to any in vivo characterization, the infectivity of SIVlhoest for macaque and human PBMC was compared to that of representative members of the SIVsm, SIVsyk, SIVagm, and HIV-1 lineage (Fig. 1). Like SIVsmE543-3, SIVlhoest infected both human and macaque PBMC efficiently. This contrasted with the restricted tropism of SIVsyk for Sykes monkey PBMC (not shown), SIVagm for macaque PBMC, and HIV-1 for human PBMC. Infectivity in macaque PBMC is a prerequisite for a robust infection in vivo but clearly is not predictive of pathogenicity for that species. To evaluate the potential pathogenicity of SIVlhoest, a cohort of four pigtailed macaques (M. nemestrina) was inoculated intravenously with uncloned SIVlhoest. Since this latter virus had been passaged twice through human T-cell lines, which might produce attenuation, we also isolated virus from a homogenate of cryopreserved spleen from the same l hoest monkey by short-term coculture with M4C8 cells (4 days) and subsequent infection of macaque PBMC. This virus was designated SIVlhoest-S to distinguish it from the PBMC isolate; four additional pigtailed macaques were inoculated intravenously with this primary virus isolate. Each of the macaques became persistently infected, as evidenced by isolation of virus from PBMC and lymph node biopsies at multiple time points postinoculation (Table 1). The macaques inoculated with the virus isolate from PBMC (macaques 622, 623, 626, and 627) that was passaged twice in human T-cell lines exhibited less-consistent virus isolation than macaques inoculated with the splenic isolate (macaques 633, 634, 635, and 636), which is suggestive of some degree of viral attenuation of the PBMC isolate. However, one of these (macaques 622) exhibited the most profound and early CD4 depletion of all the inoculated animals. Each macaque exhibited an early decline in all lymphocyte subsets (CD4 subset shown in Fig. 2A), reaching a lowest point at 1 week postinoculation. Lymphopenia was coincident with peak levels of infectious SIV in the plasma (1,000 50% tissue culture infectious doses/ml) and a high proportion of SIV-expressing cells within lymph nodes, as demonstrated by limiting dilution coculture of disrupted lymph node cells with CEMss cells (1 in 1,000 cells). High lymphoid virus expression was confirmed by ISH with SIVlhoest-specific riboprobes (Fig. 3). The plasma viremia resolved by 3 weeks, with declining prevalence of SIV-expressing cells in lymph nodes; this phase was associated with the onset of massive lymphoid hyperplasia and the trapping of virions in a pattern characteristic of the distribution of follicular dendritic cells in germinal centers (6). Subsequent fluorescenceactivated cell sorter analysis of lymphocyte subsets revealed a gradual decline (2- to 10-fold) in the numbers of circulating CD4 lymphocytes, similar to that observed in a cohort of six pigtailed macaques inoculated with a pathogenic uncloned SIVsm isolate (SIVsmE660; Fig. 2B). As was also observed with SIVsmE660 (22), the severity of the CD4 decline in

5 VOL. 73, 1999 NOVEL SIV FROM L HOEST MONKEYS 1039 FIG. 2. Characteristics of SIVlhoest infection of pigtailed macaques. (A) Kinetics of sequential alterations in CD4 lymphocytes in the peripheral blood during the first 16 weeks postinoculation in eight inoculated macaques, where the open symbols and dotted lines indicate animals inoculated with the PBMC isolate and solid lines and black symbols indicate animals inoculated with the splenic isolate. (B) Kinetics of mean CD4 lymphocyte numbers with standard deviations are compared between SIVlhoest-inoculated macaques and a cohort of six macaques inoculated with SIVsmE660, demonstrating that SIVlhoest induces a profile in declining CD4 lymphocyte numbers similar to that observed with an AIDS-inducing SIV isolate. SIVlhoest-infected macaques varied considerably, from mild to profound depletion. The macaque that exhibited the most severe CD4 depletion (macaque 622) has also failed to gain weight and exhibits mild anemia (hematocrit of 28%), thrombocytopenia ( 30,000 per l), and periodic bouts of diarrhea. Two other macaques currently exhibit mild weight loss in association with peripheral CD4 lymphocytes of less than 500 per l. SIVlhoest is a novel member of the SIVmnd lineage. In order to characterize SIVlhoest molecularly, we used degenerate primers to amplify a 120-bp fragment of the pol gene from cellular DNA extracted from infected M4C8 cells. This fragment was cloned, and based on the sequence, we designed a reverse primer in pol that was used in combination with a forward primer in the highly conserved primer binding site to amplify and clone a 2.4-kb gag-pol fragment. This fragment was used as a probe in a subsequent Southern blot hybridization to identify restriction enzymes useful for cloning and as a probe to identify proviruses within a bacteriophage lambda library generated by SstI digestion of cellular DNA extracted from infected M4C8 cells. One full-length clone, SIVlhoest-P7 was obtained and purified. SIVlhoest-P7 was infectious after transfection of M4C8 or CEMss cells and subsequent infection of macaque and human PBMC. After being subcloned into plasmid vectors, the proviral portion of the clone was sequenced in its entirety (9,957 nt) and compared to the sequences of other known primate lentiviruses. The genomic organization of SIVlhoest was similar to that of SIVagm, SIVmnd, and SIVsyk. Each of these viruses encodes gag, pol, and env, as well as the accessory genes vif, vpr, tat, rev, and nef, but lacks the additional genes vpu (found only among the members of the HIV-1 and SIVcpz lineage) and vpx (specific to SIVsm, SIVmac, and HIV-2). The long terminal repeat (LTR) of SIVlhoest (789 nt) contained all the characteristic features of other primate lentivirus LTRs, including one NF- B site and two potential SP-1 binding sites (data not shown). Comparisons of the predicted protein sequences encoded by the eight common genes revealed that SIVlhoest was quite distinct from all other SIV (and HIV) isolates analyzed to date, exhibiting at least 33% amino acid sequence difference from representatives of each of the five lineages of primate lentiviruses (Table 2). For the large genes gag, pol, and env, as well as for vpr and tat, the SIVlhoest proteins were most similar to those of SIVmnd, but for the vif, rev, and nef genes, the distance between the SIVlhoest and SIVmnd proteins was similar to that between SIVlhoest and other SIVs. The similarity between SIVlhoest and SIVmnd is shown in an alignment of the surface unit (SU) portion of the Env protein of these two viruses (Fig. 4). Although scattered substitutions are evident throughout gp120, many of the cysteine residues (asterisks) and potential N-linked glycosylation sites were conserved and regions such as the V3 loop analog and the CD4 binding domain showed remarkable conservation. The sequence of the envelope amplified from the spleen of the l hoest monkey (lhoest-s) had 95% identity with that of the infectious clone (lhoest-7). These two envelope sequences are representative of the two isolates used to inoculate the macaques. As expected from other primate lentivirus envelopes, the V1 region was the most variable, with characteristic threonine residues and insertion/deletion polymorphism. In contrast, the CD4 binding domain was absolutely conserved and only one substitution was observed in the V3 loop analog. To examine the divergence of SIVlhoest from the other primate lentiviruses in more detail, the extent of sequence difference was determined for moving windows of 200 amino acids. The resulting diversity plot confirmed that overall, SIVlhoest is most similar to SIVmnd but in some segments these two viruses are about as different from each other as they are from other SIVs (Fig. 5). Furthermore, that plot showed that the boundaries of these regions lie within genes, so that each of the major genes (gag, pol, and env) includes segments of both types. This variation in the extent of relative divergence between SIVlhoest and SIVmnd (i.e., relative to the extent of divergence from other SIVs) could indicate that the ancestors

6 1040 HIRSCH ET AL. J. VIROL. FIG. 3. Kinetics of virus expression in lymph nodes by ISH. (A1 and A2) ISH of lymph node biopsy sample obtained 1 week postinoculation, demonstrating high virus expression. Magnification, 6 (top) and 55 (bottom). (B1 and B2) Lymph node biopsy sample obtained 2 weeks after inoculation, showing a reduction in the numbers of SIV-positive cells relative to that observed at 1 week. Magnification, 6 (top) and 55 (bottom). (C1 and C2) ISH of a lymph node biopsy sample obtained 4 weeks after inoculation, showing a further reduction in the numbers of SIV-positive cells, with diffuse hybridization localized in the crescentric distribution of follicular dendritic cells within the germinal center consistent with trapping of immune complexes containing SIV on dendritic cells. Magnification, 6 (top) and 55 (bottom). of these viruses were generated by recombination of different SIV lineages, as has been found for SIVagm from sabaeus monkeys (29). To examine the evolutionary relationship of SIVlhoest to the other primate lentiviruses, we generated numerous phylogenetic trees derived from alignments of individual gene products and from smaller regions defined by consideration of the diversity plot (data not shown). These all indicated a clustering of SIVlhoest with SIVmnd, though their relative distances varied. Thus, these analyses provided no evidence that recombination has played a significant role in the evolution of SIVlhoest (or SIVmnd) and suggested that the variation in the extent of relative divergence between SIVlhoest and SIVmnd reflects changes in rates of evolution specific to these viruses. TABLE 2. Comparison of amino acid identity among primate lentiviruses, demonstrating that SIVlhoest is distantly related to SIVmnd a Comparisons of SIVlhoest and major SIV lineages Gag Pol Vif Vpr Tat Rev Env Nef SIVmnd SIVsyk SIVagm SIVsm SIVcpz a Identities were calculated as described in the Sequence comparisons section of Materials and Methods. Since the SIVlhoest-SIVmnd clade was maintained across all of the analyses, an overview of the phylogenetic relationships between these viruses was obtained from an analysis of an alignment of concatenated Gag-Pol-Env proteins, in which SIVlhoest appears as a highly divergent member of the same major lineage as SIVmnd (Fig. 6). The interpretation that SIVlhoest and SIVmnd are members of the same clade is dependent on the position of the root of the phylogenetic tree; the tree shown in Fig. 6 has been midpoint rooted. The precise position of the root of the primate lentivirus tree is problematic, since the nearest available outgroup sequences, namely, lentiviruses from other mammalian hosts, are quite distantly related. However, analyses using various such outgroups suggest that the rooting shown in Fig. 6 is appropriate. Importantly, none of our analyses using nonprimate lentivirus outgroups ever placed the root in such a position as to disrupt the SIVlhoest-SIVmnd clade. Thus, we conclude that SIVlhoest and SIVmnd are both members, albeit distantly TABLE 3. Divergence of SIVlhoest and SIVmnd from members of the African green monkey family a % Identity within lineages for indicated protein: Gag Pol Vif Vpr Tat Rev Env Nef SIVagm SIVcpz a Identities were calculated as described in the Sequence comparisons section of Materials and Methods.

7 VOL. 73, 1999 NOVEL SIV FROM L HOEST MONKEYS 1041 FIG. 4. Comparison of the predicted protein sequence of the surface subunit (SU) of the envelope of SIVlhoest and SIVmnd reveals remarkable conservation of cysteine residues and regions such as the V3 loop analog and CD4 binding domain. Conserved cysteines are indicated by, and variable cysteine residues are indicated by a above the top sequence. Potential N-linked glycosylation sites are underlined. The predicted sequence of gp120 of the SIVlhoest-7 molecularly cloned virus derived from a PBMC isolate is shown on the top (lhoest-7). Substitutions relative to this sequence in the predicted sequence of a clone of envelope amplified directly from the spleen of this monkey (lhoest-s) and the SIVmnd/GB-1 clone are shown aligned below. Dots indicate amino acid identity at a residue, and a dash indicates a gap introduced to optimize alignment. Variable regions analagous to those observed in HIV-1 and other SIVs are indicated, and the cleavage site for the transmembrane glycoprotein (TM) is shown. related, of the same major primate lentivirus lineage. Overall, SIVlhoest and SIVmnd are rather more divergent from each other than are SIVs from different species of African green monkey (Table 3). The only other example of such high divergence within a major lineage involves the SIVcpz-HIV-1 lineage, where SIVcpz/Ant is almost as different from SIVcpz/Gab1 (Table 3) and HIV-1 as SIVlhoest and SIVmnd are from each other. DISCUSSION It seems clear that HIV-1 originated through cross-species transmission(s) in the recent past from a naturally infected African primate, but the species involved remains open to question. Therefore, further exploration of the nature of the diversity among primate lentiviruses is necessary to elucidate the origins and evolution of the human viruses. Now that we have characterized SIVlhoest and examined the relationship between it and the previously known primate lentiviruses, it is clear that this particular virus is not the proximal source of HIV-1. However, the phylogenetic position of SIVlhoest is surprising and has implications for our understanding of primate lentivirus evolution. In particular, at first sight, the genetic similarity between SIVlhoest and SIVmnd seems difficult to explain. While the phylogeny of the primate lentiviruses indicates that there have been multiple cross-species transmissions (45), there are also indications that some of the viruses have co-

8 1042 HIRSCH ET AL. J. VIROL. FIG. 5. Diversity plot comparing SIVlhoest with representatives of each of the five major lineages of primate lentiviruses, i.e., SIVmnd, SIVsyk, SIVsm, SIVcpz, and SIVagm (SIVver). Protein sequence difference is plotted for windows of 200 amino acids moved in steps of 10. FIG. 6. Phylogenetic relationship of SIVlhoest to other primate lentiviruses. The tree was derived by maximum likelihood analysis of a concatenated Gag- Pol-Env protein alignment (see text for details). A tree derived by neighborjoining analysis differed in no significant way. Stars indicate that the clade to the right was found in 100% of bootstrap replicates of the neighbor-joining analysis. Horizontal branch lengths are drawn to scale, with the bar indicating 0.1 amino acid replacement per site. evolved with their natural host species and thus that the group as a whole may be quite ancient. The four species of African green monkeys (vervets, grivets, and sabaeus and tantalus monkeys) each harbor their own variants of SIVagm, while SIVsyk and now SIVlhoest are rather more divergent viruses from rather more divergent Cercopithecus species. It is tempting to speculate that these Cercopithecus SIVs represent the vertical transmission backbone of the primate lentivirus phylogeny, with the other SIVs from more distantly related primates having all resulted from horizontal transmissions. Whether this scenario is accurate, the comparatively close relationship between SIVlhoest and SIVmnd seems most unlikely to be a reflection of host-dependent viral evolution, since the mandrill belongs to a genus (Mandrillus) quite distant from the Cercopithecus genus, and so (at least) one of these viruses is likely the result of cross-species transmission. There is accumulating evidence that such transfer of SIV does occur naturally between different species of monkeys that share a common habitat in the wild. For example, the sabaeus subtype of SIVagm has been identified in wild-caught patas monkeys (Erythorocebus patas) in West Africa (5) and the vervet subtype of SIVagm has been found in a yellow baboon (Papio hamadryas cynocephalus) in Tanzania (29). However, mandrills are restricted to the west coastal region of central Africa around Gabon, whereas l hoest monkeys inhabit a localized area of central Africa more than 1,000 km to the east (Fig. 7) (32, 34, 42). The geographical separation of l hoest monkeys and mandrills implies that neither could be the immediate source of virus for the other. However, close relatives of l hoest monkeys are found in regions of west central Africa overlapping the mandrill range (Fig. 7). Preussis monkeys (C. preussi preussi

9 VOL. 73, 1999 NOVEL SIV FROM L HOEST MONKEYS 1043 FIG. 7. A schematic view of Africa, showing the ranges occupied by l hoest monkeys (C. l hoesti l hoesti) and their close relatives, preussis monkeys (C. preussi preussi) and suntailed monkeys (C. solatus); the distribution of mandrills (Mandrillus sphinx) is indicated by cross-hatching. A representative l hoest monkey is shown. and insularis) from Cameroon and Bioko Island and sun-tailed monkeys (C. solatus) from Gabon are sufficiently similar to l hoest monkeys that they have been considered by some to be a subspecies of C. lhoesti. Furthermore, at least one of these species, C. solatus, has serologic evidence of SIV infection (25a). We therefore hypothesize that SIVmnd infection of mandrills resulted from cross-species transmission of SIV from one of these relatives of the l hoest monkey. This direction of the transmission, rather than from mandrill to the l hoest monkey, is consistent with the Cercopithecus origin of primate lentiviruses suggested above and is supported by the apparently low rate of SIV seroprevalence in wild mandrills. Only two seropositive wild-caught mandrills have been reported, and the only existing clone came from one of two seropositive founders of a colony in Gabon. Although SIVmnd is now prevalent within this colony, all of the circulating viruses appear to be highly related, suggesting subsequent transmission of this virus within the colony (16, 39). It will be interesting to determine whether feral mandrills from other locations also harbor SIVmnd. Consistent with the proposed ancient relationship between primate lentiviruses and their respective natural hosts, SIVs appear to result in an asymptomatic infection in their natural host species (e.g., SIVsm in sooty mangabeys or SIVagm in African green monkeys) (24). However, AIDS may result upon experimental infection of other primates, particularly macaques (2, 7, 22, 24, 35). In the present study, SIVlhoestinfected macaques demonstrated many of the virologic, immunologic, and clinical characteristics of early infection of macaques with pathogenic isolates of SIVsm, SIVmac, or SIVagm. Such characteristics included (i) high viral expression in lymphoid tissues in the primary phase of infection, (ii) high primary plasma viremia, (iii) subsequent lymphadenopathy and trapping of viral RNA in germinal centers in a follicular dendritic cell pattern, (iv) declining peripheral CD4 lymphocyte numbers, (v) persistent PBMC-associated viremia, and (vi) thrombocytopenia and weight loss in one macaque. Although these animals have not been infected for a sufficient period of time to become symptomatic, the virologic and clinical features of infection are consistent with progression to AIDS. Moreover, SIVlhoest appears to be directly pathogenic for macaques without adaptation by prior macaque passage. The ability of SIVlhoest to infect human PBMC in vitro at least as efficiently as it infects macaque PBMC suggests that this virus has the potential to infect human populations. Such cross-species transmission to humans has already been observed for the SIVsm isolate that is now circulating among human populations as HIV-2 (13, 14). While the ability of this virus to infect human PBMC may not be predictive of virulence in humans, serologic surveys of humans in regions near the habitat of l hoest monkeys are necessary to evaluate this possibility. These highly specialized forest dwellers have considerably less contact with humans than species such as sooty mangabeys. However, the continued expansion of human populations and encroachment upon the habitats of primates such as the l hoest monkey may amplify the risk for such an event in the future.

10 1044 HIRSCH ET AL. J. VIROL. ACKNOWLEDGMENTS We thank Russell Byrum and Marisa St. Claire, Bioqual, Inc., Rockville, Md., for assistance in conducting the animal studies; Michael Durham, Oregon Zoo, Portland, Ore., for providing pictures of l hoest monkeys; and John Oates, Hunter College, New York, N.Y., for useful discussions of primatology. REFERENCES 1. Adachi, J., and M. Hasegawa MOLPHY (a program package for MOLecular PHYlogenetics), version 2.2. Institute of Statistical Mathematics, Tokyo, Japan. 2. Allan, J. S Pathogenic properties of simian immunodeficiency viruses in nonhuman primates. Annu. Rev. AIDS Res 1: Allan, J. S., M. Short, M. E. Taylor, S. Su, V. M. Hirsch, P. R. Johnson, G. M. Shaw, and B. H. Hahn Natural infection of West African green monkeys with a unique subtype of simian immunodeficiency virus SIVagm. J. Virol. 65: Baier, M., A. Werner, K. Cichutek, C. Garber, C. Muller, G. Kraus, F. J. Ferdinand, S. Hartung, T. S. Papas, and R. Kurth Molecularly cloned simian immunodeficiency virus SIVagm3 is highly divergent from other SIVagm isolates and is biologically active in vitro and in vivo. J. Virol. 63: Bibollet-Ruche, F., A. Galat-Luong, G. Cuny, P. Sarni-Manchado, G. Galat, J. P. Durant, X. Pourrut, and F. Veas Simian immunodeficiency virus infection in a patas monkey (Erythrocebus patas): evidence for cross-species transmission from African green monkeys (Cercopthecus aethiops sabeus) in the wild. J. Gen. Virol. 77: Chalifoux, L. V., D. J. Ringler, N. W. King, P. K. Seghal, R. C. Desrosiers, M. D. Daniel, and N. L. Letvin Lymphadenopathy in macaques experimentally infected with simian immunodeficiency virus (SIV). Am. J. Pathol. 128: Chen, Z., P. Telfer, P. Reed, A. Gettie, L. Zhand, D. D. Ho, and P. Marx Genetic characterization of new West African simian immunodeficiency virus SIVsm: geographic clustering of household-derived SIV strains with human immunodeficiency virus type 2 subtypes and genetically diverse viruses from a single feral sooty mangabey troop. J. Virol. 70: Daniel, M. D., Y. Li, Y. M. Naidu, P. J. Durda, D. K. Schmidt, C. D. Troup, D. P. Silva, J. J. MacKey, H. W. Kestler, P. K. Seghal, et al Simian immunodeficiency viruses from African green monkeys. J. Virol. 62: Emau, P., H. M. McClure, M. Isahakai, J. G. Else, and P. N. Fultz Isolation from African Sykes monkeys (Cercopithecus mitis) of a lentivirus related to human and simian immunodeficiency viruses. J. Virol. 65: Fomsgaard, A., V. M. Hirsch, J. S. Allan, and P. R. Johnson A highly divergent proviral DNA clone of SIV from a distinct species of African green monkey. Virology 182: Fukasawa, M., T. Miura, A. Hasegawa, S. Morikawa, H. Tsuijmoto, K. Miki, T. Kitamura, and M. Hayami Sequence of simian immunodeficiency virus from African green monkey, a new member of the HIV/SIV group. Nature 333: Galtier, N., M. Gouy, and C. Gautier SEAVIEW and PHYLO_WIN: two graphic tools for sequence alignment and molecular phylogeny. Comp. Appl. Biosci. 12: Gao, F. L., L. Yue, A. T. White, P. G. Pappas, J. Barchue, A. J. Hanson, B. M. Green, P. M. Sharp, G. M. Shaw, and B. H. Hahn Human infection by genetically diverse SIVsm-related HIV-2 in West Africa. Nature 358: Gao F. L., L. Yue, A. T. White, P. G. Pappas, J. Barchue, A. J. Hanson, B. M. Green, P. M. Sharp, G. M. Shaw, and B. H. Hahn Genetic diversity of human immunodeficiency viruses type 2: evidence for distinct sequence subtypes with differences in virus biology. J. Virol. 68: Gelman, I. H., J. Zhang, E. Hailman, H. Hanafusa, and S. S. Morse Identification and evaluation of new primer sets for the detection of lentivirus proviral DNA. AIDS Res. Hum. Retroviruses 8: Georges-Courbot, M. C., P. Moisson, E. Leroy, A. M. Pingard, E. Nerrienet, G. Dubreuil, E. J. Wickings, F. Debels, I. Bedjabaga, V. Poaty-Mavoungou, N. T. Hahn, and A. J. Georges Occurrence and frequency of transmission of naturally occurring simian retroviral infections (SIV, STLV and SRV) at the CIRMF primate center, Gabon. J. Med. Primatol. 25: Georges-Courbot, M. C., C. Y. Lu, M. Makuwa, P. Telfer, R. Onanga, G. Dubreuil, Z. Chen, S. M. Smith, A. Georges, F. Gao, B. H. Hahn, and P. A. Marx Natural infection of a household pet red-capped mangabey (Cercocebus torquatus torquatus) with a new simian immunodeficiency virus. J. Virol. 72: Hendry, R. M., M. A. Wells, M. A. Phelan, A. L. Schneider, J. S. Epstein, and G. V. Quinnan Antibodies to simian immunodeficiency virus in African green monkeys in Africa in Lancet ii: Hirsch, V. M., R. A. Olmsted, M. Murphey-Corb, R. H. Purcell, and P. R. Johnson An African primate lentivirus (SIVsm) closely related to HIV-2. Nature 339: Hirsch, V. M., C. McGann, G. Dapolito, S. Goldstein, A. Ogen-Odoi, B. Biryawaho, T. Lakwo, and P. R. Johnson Identification of a new subgroup of SIVagm in tantalus monkeys. Virology 197: Hirsch, V. M., G. Dapolito, S. Goldstein, H. McClure, P. Emau, P. N. Fultz, M. Isahakia, R. Neroot, G. Myers, and P. R. Johnson A distinct African lentivirus from Sykes monkeys. J. Virol. 67: Hirsch, V. M., and P. R. Johnson Genetic diversity and phylogeny of primate lentiviruses, p In J. Morrow and N. Haigwood (ed.), HIV molecular organization, pathogenicity and treatment. Elsevier Science Publishers, Amsterdam, The Netherlands. 23. Hirsch, V. M., and P. R. Johnson Pathogenic diversity of simian immunodeficiency viruses. Virus Res. 32: Hirsch, V. M., G. Dapolito, P. R. Johnson, W. R. Elkins, W. T. London, R. J. Montali, S. Goldstein, and C. Brown Induction of AIDS by simian immunodeficiency virus from an African green monkey: species-specific variation in pathogenicity correlates with the extent of in vivo replication. J. Virol. 69: Hirsch, V. M., G. Dapolito, R. Goeken, and B. Campbell Phylogeny and natural history of the primate lentiviruses, SIV and HIV. Curr. Opin. Genet. Dev. 5: a.Hirsch, V. M. Unpublished data. 26. Huet, T., R. Cheynier, A. Meyerhans, G. Roelants, and S. Wain-Hobson Genetic organization of a chimpanzee lentivirus related to HIV-1. Nature 345: Janssens, W., K. Fransen, M. Peeters, L. Heyndrickx, J. Motte, L. Bedjabaga, E. Depaporte, P. Piot, and G. Van Der Groen Phylogenetic analysis of a new chimpanzee lentivirus SIVcpz-gab-2 from a wild-captured chimpanzee from Gabon. AIDS Res. Hum. Retroviruses 10: Jin, M. J., J. Rogers, J. E. Phillips-Conroy, J. S. Allan, R. C. Desrosiers, G. M. Shaw, P. M. Sharp, and B. H. Hahn Mosaic genome structure of simian immunodeficiency virus from West African green monkeys. EMBO J. 13: Jin, M. J., J. Rogers, J. E. Phillips-Conroy, J. S. Allan, R. C. Desrosiers, G. M. Shaw, P. M. Sharp, and B. H. Hahn Infection of a yellow baboon with simian immunodeficiency virus from African green monkeys: evidence for cross-species transmission in the wild. J. Virol. 68: Johnson, P. R., A. Fomsgaard, J. Allan, M. Gravell, W. T. London, R. A. Olmsted, and V. M. Hirsch Simian immunodeficiency viruses from African green monkeys display unusual genetic diversity. J. Virol. 64: Jones, D. T., W. R. Taylor, and J. M. Thornton The rapid generation of mutation data matrices from protein sequences. Comp. Appl. Biosci. 8: Kingdon, J East African mammals: an atlas of evolution in Africa, vol. 1. The University of Chicago Press, Chicago, Ill. 33. Kodama, T., D. P. Silva, M. D. Daniel, J. E. Phillips-Conroy, C. J. Jolly, J. Rogers, and R. C. Desrosiers Prevalence of antibodies to SIV in baboons in their native habitat. AIDS Res. Hum. Retroviruses 5: Lernould, J.-M Classification and geographical distribution of guenons: a review, p In A. Gautier-Hion, F. Bouliere, C. Gautier, and J. Kingdon (ed.), A primate radiation: evolutionary biology of the African guenons. Cambridge University Press, Cambridge, United Kingdom. 35. Letvin, N. L., and N. W. King Immunologic and pathologic manifestations of the infection of rhesus monkeys with simian immunodeficiency virus of macaques. J. Acquired Immune Defic. Syndr. 3: Lowenstine, L. J., N. C. Pederson, J. Higgins, K. C. Pallis, A. Uyeda, P. Marx, N. W. Lerche, R. J. Munn, and M. B. Gardner Seroepidemiologic survey of captive Old-World primates for antibodies to human and simian retroviruses, and isolation of a lentivirus from a sooty mangabeys (Cercocebus atys). Int. J. Cancer 38: Marx, P., Y. Li, N. W. Lerche, S. Sutjipto, A. Gettie, J. A. Yee, B. Brotman, A. M. Prince, A. Hanson, R. G. Webster, and R. C. Desrosiers Isolation of a simian immunodeficiency virus related to human immunodeficiency virus type 2 from a West African pet sooty mangabey. J. Virol. 65: Muller, M. C., N. K. Saksena, E. Nerrienet, C. Chappey, V. M. Herve, J. P. Durand, P. Legal-Campodonico, M. C. Lang, J. P. Digoutte, A. J. Georges, P. Sonigo, and F. Barre-Sinoussi Simian immunodeficiency viruses from central and western Africa: evidence for a new species-specific lentivirus in tantalus monkeys. J. Virol. 67: Nerrienet, E., X. Amouretti, M. C. Muller-Trutwin, V. Poaty-Mavoungou, I. Bedjebaga, H. T. Ngyuyen, G. Dubreuil, S. Corbet, E. J. Wickings, F. Barre- Sinoussi, A. J. Georges, and M.-C. Georges-Corbot Phylogenetic analysis of SIV and STLV-1 in mandrills (Mandrillus sphinx): indications that intracolony transmissions are predominantly the result of male-to-male aggressive contacts. AIDS Res. Hum. Retroviruses 14: Nicol, I., D. Messinger, P. Dibouch, J. Bernard, I. Desportes, R. Jouffre, R. Snart, P. Nara, R. C. Gallo, and D. Zagury Use of Old World monkeys for acquired immunodeficiency syndrome research. J. Med. Primatol. 18: Oates, J. F The distribution of Cercopithecus monkeys in West African forests, p In A. Gautier-Hion, F. Bouliere, C. Gautier, and J. Kingdon (ed.), A primate radiation: evolutionary biology of the African

JOURNAL OF VIROLOGY, Feb. 1999, p Vol. 73, No. 2. Copyright 1999, American Society for Microbiology. All Rights Reserved.

JOURNAL OF VIROLOGY, Feb. 1999, p Vol. 73, No. 2. Copyright 1999, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Feb. 1999, p. 1036 1045 Vol. 73, No. 2 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Characterization of a Novel Simian Immunodeficiency

More information

Simian immunodeficiency viruses (SIVs) from eastern and southern Africa: detection of a SIVagm variant from a chacma baboon

Simian immunodeficiency viruses (SIVs) from eastern and southern Africa: detection of a SIVagm variant from a chacma baboon Journal of General Virology (1998), 79, 1809 1814. Printed in Great Britain.... SHORT COMMUNICATION Simian immunodeficiency viruses (SIVs) from eastern and southern Africa: detection of a SIVagm variant

More information

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES 1 of 7 I. Viral Origin. A. Retrovirus - animal lentiviruses. HIV - BASIC PROPERTIES 1. HIV is a member of the Retrovirus family and more specifically it is a member of the Lentivirus genus of this family.

More information

Wide Range of Viral Load in Healthy African Green Monkeys Naturally Infected with Simian Immunodeficiency Virus

Wide Range of Viral Load in Healthy African Green Monkeys Naturally Infected with Simian Immunodeficiency Virus JOURNAL OF VIROLOGY, Dec. 2000, p. 11744 11753 Vol. 74, No. 24 0022-538X/00/$04.00 0 Wide Range of Viral Load in Healthy African Green Monkeys Naturally Infected with Simian Immunodeficiency Virus SIMOY

More information

Risks and Management of SIV in De Brazza Guenon (Cercopithecus neglectus)

Risks and Management of SIV in De Brazza Guenon (Cercopithecus neglectus) Risks and Management of SIV in De Brazza Guenon (Cercopithecus neglectus) NB: This paper is produced specifically for SIV in De Brazza Monkeys. There are significant species differences and therefore the

More information

Wild Mandrillus sphinx Are Carriers of Two Types of Lentivirus

Wild Mandrillus sphinx Are Carriers of Two Types of Lentivirus JOURNAL OF VIROLOGY, Aug. 2001, p. 7086 7096 Vol. 75, No. 15 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.15.7086 7096.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Wild Mandrillus

More information

Received 19 July 2002/Accepted 24 September 2002

Received 19 July 2002/Accepted 24 September 2002 JOURNAL OF VIROLOGY, Jan. 2003, p. 744 748 Vol. 77, No. 1 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.1.744 748.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Partial Molecular

More information

A novel simian immunodeficiency virus from black mangabey (Lophocebus aterrimus) in the Democratic Republic of Congo

A novel simian immunodeficiency virus from black mangabey (Lophocebus aterrimus) in the Democratic Republic of Congo Journal of General Virology (2005), 86, 1967 1971 DOI 10.1099/vir.0.80697-0 Short Communication A novel simian immunodeficiency virus from black mangabey (Lophocebus aterrimus) in the Democratic Republic

More information

Natural Infection of a Household Pet Red-Capped Mangabey (Cercocebus torquatus torquatus) with a New Simian Immunodeficiency Virus

Natural Infection of a Household Pet Red-Capped Mangabey (Cercocebus torquatus torquatus) with a New Simian Immunodeficiency Virus JOURNAL OF VIROLOGY, Jan. 1998, p. 600 608 Vol. 72, No. 1 0022-538X/98/$04.00 0 Copyright 1998, American Society for Microbiology Natural Infection of a Household Pet Red-Capped Mangabey (Cercocebus torquatus

More information

Risks and Management of SIV in Mandrils (Mandrillus sphinx) and Drills (Mandrillus leucophaeus)

Risks and Management of SIV in Mandrils (Mandrillus sphinx) and Drills (Mandrillus leucophaeus) Risks and Management of SIV in Mandrils (Mandrillus sphinx) and Drills (Mandrillus leucophaeus) A. Background Simian Immunodeficiency Viruses (SIV) are primate lentiviruses, which infect a wide variety

More information

Received 30 January 2002/Accepted 10 May 2002

Received 30 January 2002/Accepted 10 May 2002 JOURNAL OF VIROLOGY, Aug. 2002, p. 8298 8309 Vol. 76, No. 16 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.16.8298 8309.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. Characterization

More information

OPEN ACCESS JZAR Evidence-based practice. Evidence-based practice. Introduction. Methods. M.P. Hartley 1,* and F. Schmidt 2

OPEN ACCESS JZAR Evidence-based practice. Evidence-based practice. Introduction. Methods. M.P. Hartley 1,* and F. Schmidt 2 OPEN ACCESS JZAR Evidence-based practice Evidence-based practice The use of risk analysis methodology to generate evidence-based decision making in zoo animal disease management: using simian immunodeficiency

More information

Micro 301 HIV/AIDS. Since its discovery 31 years ago 12/3/ Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people

Micro 301 HIV/AIDS. Since its discovery 31 years ago 12/3/ Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people Micro 301 HIV/AIDS Shiu-Lok Hu hus@uw.edu December 3, 2012 Since its discovery 31 years ago Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people In 2011 34.0 million [31.4 35.9 million]

More information

reproduced or photocopying

reproduced or photocopying AIDS Reviews 2004;6:40-53 What Can Natural Infection of African Monkeys with Simian Immunodeficiency Virus Tell us About the Pathogenesis of AIDS? Vanessa M. Hirsch Laboratory of Molecular Microbiology,

More information

An Evolutionary Story about HIV

An Evolutionary Story about HIV An Evolutionary Story about HIV Charles Goodnight University of Vermont Based on Freeman and Herron Evolutionary Analysis The Aids Epidemic HIV has infected 60 million people. 1/3 have died so far Worst

More information

Mosaic Genomes of the Six Major Primate Lentivirus Lineages Revealed by Phylogenetic Analyses

Mosaic Genomes of the Six Major Primate Lentivirus Lineages Revealed by Phylogenetic Analyses REFERENCES CONTENT ALERTS Mosaic Genomes of the Six Major Primate Lentivirus Lineages Revealed by Phylogenetic Analyses Marco Salemi, Tulio De Oliveira, Valerie Courgnaud, Vincent Moulton, Barbara Holland,

More information

Lentiviruses: HIV-1 Pathogenesis

Lentiviruses: HIV-1 Pathogenesis Lentiviruses: HIV-1 Pathogenesis Human Immunodeficiency Virus, HIV, computer graphic by Russell Kightley Tsafi Pe ery, Ph.D. Departments of Medicine and Biochemistry & Molecular Biology NJMS, UMDNJ. e-mail:

More information

Received 29 March 1999/Accepted 17 June 1999

Received 29 March 1999/Accepted 17 June 1999 JOURNAL OF VIROLOGY, Oct. 1999, p. 7994 8002 Vol. 73, No. 10 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Discovery of a New Endogenous Type C Retrovirus

More information

Fayth K. Yoshimura, Ph.D. September 7, of 7 RETROVIRUSES. 2. HTLV-II causes hairy T-cell leukemia

Fayth K. Yoshimura, Ph.D. September 7, of 7 RETROVIRUSES. 2. HTLV-II causes hairy T-cell leukemia 1 of 7 I. Diseases Caused by Retroviruses RETROVIRUSES A. Human retroviruses that cause cancers 1. HTLV-I causes adult T-cell leukemia and tropical spastic paraparesis 2. HTLV-II causes hairy T-cell leukemia

More information

Going Nowhere Fast: Lentivirus genetic sequence evolution does not correlate with phenotypic evolution.

Going Nowhere Fast: Lentivirus genetic sequence evolution does not correlate with phenotypic evolution. Going Nowhere Fast: Lentivirus genetic sequence evolution does not correlate with phenotypic evolution. Brian T. Foley, PhD btf@lanl.gov HIV Genetic Sequences, Immunology, Drug Resistance and Vaccine Trials

More information

Did Africans get HIV from chimps?

Did Africans get HIV from chimps? VOLUME 7, NUMBER 8 RETHINKING AIDS www.rethinkingaids.com AUGUST 1999 Did Africans get HIV from chimps? Not likely, asserts biophysicist Eleni Papadopulos-Eleopulos in her latest paper rejected by Nature

More information

Isolation from African Sykes' Monkeys (Cercopithecus mitis) of a Lentivirus Related to Human and Simian Immunodeficiency Viruses

Isolation from African Sykes' Monkeys (Cercopithecus mitis) of a Lentivirus Related to Human and Simian Immunodeficiency Viruses JOURNAL OF VIROLOGY, Apr. 1991, p. 2135-2140 0022-538X/91/042135-06$02.00/0 Copyright C) 1991, American Society for Microbiology Vol. 65, No. 4 Isolation from African Sykes' Monkeys (Cercopithecus mitis)

More information

INTRODUCTION. Gabon. Gabon

INTRODUCTION. Gabon. Gabon Journal of General Virology (2009), 90, 488 499 DOI 10.1099/vir.0.005181-0 Simian immunodeficiency virus types 1 and 2 (SIV mnd 1 and 2) have different pathogenic potentials in rhesus macaques upon experimental

More information

Viral Vectors In The Research Laboratory: Just How Safe Are They? Dawn P. Wooley, Ph.D., SM(NRM), RBP, CBSP

Viral Vectors In The Research Laboratory: Just How Safe Are They? Dawn P. Wooley, Ph.D., SM(NRM), RBP, CBSP Viral Vectors In The Research Laboratory: Just How Safe Are They? Dawn P. Wooley, Ph.D., SM(NRM), RBP, CBSP 1 Learning Objectives Recognize hazards associated with viral vectors in research and animal

More information

Micropathology Ltd. University of Warwick Science Park, Venture Centre, Sir William Lyons Road, Coventry CV4 7EZ

Micropathology Ltd. University of Warwick Science Park, Venture Centre, Sir William Lyons Road, Coventry CV4 7EZ www.micropathology.com info@micropathology.com Micropathology Ltd Tel 24hrs: +44 (0) 24-76 323222 Fax / Ans: +44 (0) 24-76 - 323333 University of Warwick Science Park, Venture Centre, Sir William Lyons

More information

Immunodeficiency Virus Type 2 from a West African

Immunodeficiency Virus Type 2 from a West African JOURNAL OF VIROLOGY, Aug. 1991, p. 448-4485 22-58X/91/8448-6$2./ Copyright 1991, American Society for Microbiology Vol. 65, No. 8 Isolation of a Simian Immunodeficiency Virus Related to Human Immunodeficiency

More information

CONCISE COMMUNICATION

CONCISE COMMUNICATION 1791 CONCISE COMMUNICATION Detection of Diverse Variants of Human Immunodeficiency Virus 1 Groups M, N, and O and Simian Immunodeficiency Viruses from Chimpanzees by Using Generic pol and env Primer Pairs

More information

The use of nonhuman primates in biomedical research has led to the isolation of many

The use of nonhuman primates in biomedical research has led to the isolation of many JVI Accepts, published online ahead of print on 29 September 2010 J. Virol. doi:10.1128/jvi.01928-10 Copyright 2010, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights

More information

SUPPLEMENTARY INFORMATION. Divergent TLR7/9 signaling and type I interferon production distinguish

SUPPLEMENTARY INFORMATION. Divergent TLR7/9 signaling and type I interferon production distinguish SUPPLEMENTARY INFOATION Divergent TLR7/9 signaling and type I interferon production distinguish pathogenic and non-pathogenic AIDS-virus infections Judith N. Mandl, Ashley P. Barry, Thomas H. Vanderford,

More information

Analysis of the Evolutionary Relationships of HIV-1 and SIVcpz Sequences Using Bayesian Inference: Implications for the Origin of HIV-1

Analysis of the Evolutionary Relationships of HIV-1 and SIVcpz Sequences Using Bayesian Inference: Implications for the Origin of HIV-1 Analysis of the Evolutionary Relationships of HIV-1 and SIVcpz Sequences Using Bayesian Inference: Implications for the Origin of HIV-1 D. Paraskevis,* P. Lemey,* M. Salemi,* M. Suchard, Y. Van de Peer,à

More information

Retroviruses. ---The name retrovirus comes from the enzyme, reverse transcriptase.

Retroviruses. ---The name retrovirus comes from the enzyme, reverse transcriptase. Retroviruses ---The name retrovirus comes from the enzyme, reverse transcriptase. ---Reverse transcriptase (RT) converts the RNA genome present in the virus particle into DNA. ---RT discovered in 1970.

More information

Human Immunodeficiency Virus

Human Immunodeficiency Virus Human Immunodeficiency Virus Virion Genome Genes and proteins Viruses and hosts Diseases Distinctive characteristics Viruses and hosts Lentivirus from Latin lentis (slow), for slow progression of disease

More information

Case Study. The origins and evolution of HIV STUDENT S GUIDE. Anne Fischer. Dean Madden [Ed.] Version 1.2

Case Study. The origins and evolution of HIV STUDENT S GUIDE. Anne Fischer. Dean Madden [Ed.] Version 1.2 STUDENT S GUIDE Case Study The origins and evolution of HIV Version 1.2 Anne Fischer Formerly of the Max Planck Institute for Evolutionary Anthropology, Leipzig Dean Madden [Ed.] NCBE, University of Reading

More information

Received 27 November 1995/Accepted 26 February 1996

Received 27 November 1995/Accepted 26 February 1996 JOURNAL OF VIROLOGY, June 1996, p. 3617 3627 Vol. 70, No. 6 0022-538X/96/$04.00 0 Copyright 1996, American Society for Microbiology Genetic Characterization of New West African Simian Immunodeficiency

More information

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection Anne Piantadosi 1,2[, Bhavna Chohan 1,2[, Vrasha Chohan 3, R. Scott McClelland 3,4,5, Julie Overbaugh 1,2* 1 Division of Human

More information

HIV INFECTION: An Overview

HIV INFECTION: An Overview HIV INFECTION: An Overview UNIVERSITY OF PAPUA NEW GUINEA SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY & MOLECULAR BIOLOGY PBL MBBS II SEMINAR VJ

More information

Received 8 October 1997/Accepted 5 January 1998

Received 8 October 1997/Accepted 5 January 1998 JOURNAL OF VIROLOGY, Apr. 1998, p. 3248 3258 Vol. 72, No. 4 0022-538X/98/$04.00 0 Copyright 1998, American Society for Microbiology In Vivo Replication Capacity Rather Than In Vitro Macrophage Tropism

More information

Received 19 March 2002/Accepted 16 July 2002

Received 19 March 2002/Accepted 16 July 2002 JOURNAL OF VIROLOGY, Oct. 2002, p. 10256 10263 Vol. 76, No. 20 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.20.10256 10263.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. High

More information

Amplification of a Complete Simian Immunodeficiency Virus Genome from Fecal RNA of a Wild Chimpanzee

Amplification of a Complete Simian Immunodeficiency Virus Genome from Fecal RNA of a Wild Chimpanzee JOURNAL OF VIROLOGY, Feb. 2003, p. 2233 2242 Vol. 77, No. 3 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.3.2233 2242.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Amplification

More information

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR Supplemental Materials and Methods Plasmids and viruses To generate pseudotyped viruses, the previously described recombinant plasmids pnl4-3-δnef-gfp or pnl4-3-δ6-drgfp and a vector expressing HIV-1 X4

More information

Human Immunodeficiency Virus. Acquired Immune Deficiency Syndrome AIDS

Human Immunodeficiency Virus. Acquired Immune Deficiency Syndrome AIDS Human Immunodeficiency Virus Acquired Immune Deficiency Syndrome AIDS Sudden outbreak in USA of opportunistic infections and cancers in young men in 1981 Pneumocystis carinii pneumonia (PCP), Kaposi s

More information

HIV/AIDS. Biology of HIV. Research Feature. Related Links. See Also

HIV/AIDS. Biology of HIV. Research Feature. Related Links. See Also 6/1/2011 Biology of HIV Biology of HIV HIV belongs to a class of viruses known as retroviruses. Retroviruses are viruses that contain RNA (ribonucleic acid) as their genetic material. After infecting a

More information

30 Years with AIDS: Where it Came From and Why It s Still With Us. Kevin McCarthy Ben Morris Jamie Schafer

30 Years with AIDS: Where it Came From and Why It s Still With Us. Kevin McCarthy Ben Morris Jamie Schafer 30 Years with AIDS: Where it Came From and Why It s Still With Us Kevin McCarthy Ben Morris Jamie Schafer 30 Years with AIDS: Where it Came From and Why It s Still With Us Kevin McCarthy Part I: The Origins

More information

DC-SIGN from African Green Monkeys Is Expressed in Lymph Nodes and Mediates Infection in trans of Simian Immunodeficiency Virus SIVagm

DC-SIGN from African Green Monkeys Is Expressed in Lymph Nodes and Mediates Infection in trans of Simian Immunodeficiency Virus SIVagm JOURNAL OF VIROLOGY, Jan. 2004, p. 798 810 Vol. 78, No. 2 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.2.798 810.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. DC-SIGN from African

More information

Department of Animal and Poultry Sciences October 16, Avian Leukosis Virus Subgroup J. Héctor L. Santiago ABSTRACT

Department of Animal and Poultry Sciences October 16, Avian Leukosis Virus Subgroup J. Héctor L. Santiago ABSTRACT Department of Animal and Poultry Sciences October 16, 2000 Avian Leukosis Virus Subgroup J Héctor L. Santiago ABSTRACT The avian leukosis viruses (ALV) are a class of retroviruses belonging to the avian

More information

CD4-Like Immunological Function by CD4 T Cells in Multiple Natural Hosts of Simian Immunodeficiency Virus

CD4-Like Immunological Function by CD4 T Cells in Multiple Natural Hosts of Simian Immunodeficiency Virus JOURNAL OF VIROLOGY, Sept. 2011, p. 8702 8708 Vol. 85, No. 17 0022-538X/11/$12.00 doi:10.1128/jvi.00332-11 Copyright 2011, American Society for Microbiology. All Rights Reserved. CD4-Like Immunological

More information

Received 24 May 1999/Accepted 22 September 1999

Received 24 May 1999/Accepted 22 September 1999 JOURNAL OF VIROLOGY, Jan. 2000, p. 529 534 Vol. 74, No. 1 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. env Sequences of Simian Immunodeficiency Viruses

More information

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute Understanding HIV Transmission Utilizing Transmitted/Founder Viruses Brandon Keele SAIC-Frederick National Cancer Institute AIDS Vaccine 2011 15 September 2011 Overview Several years ago, the CHAVI sought

More information

Received 13 August 1992/Accepted 3 December 1992

Received 13 August 1992/Accepted 3 December 1992 JOURNAL OF VIROLOGY, Mar. 1993, P. 1227-1235 0022-538X/93/031227-09$02.00/0 Copyright ) 1993, American Society for Microbiology Vol. 67, No. 3 Simian Immunodeficiency Viruses from Central and Western Africa:

More information

HOST-PATHOGEN CO-EVOLUTION THROUGH HIV-1 WHOLE GENOME ANALYSIS

HOST-PATHOGEN CO-EVOLUTION THROUGH HIV-1 WHOLE GENOME ANALYSIS HOST-PATHOGEN CO-EVOLUTION THROUGH HIV-1 WHOLE GENOME ANALYSIS Somda&a Sinha Indian Institute of Science, Education & Research Mohali, INDIA International Visiting Research Fellow, Peter Wall Institute

More information

MedChem 401~ Retroviridae. Retroviridae

MedChem 401~ Retroviridae. Retroviridae MedChem 401~ Retroviridae Retroviruses plus-sense RNA genome (!8-10 kb) protein capsid lipid envelop envelope glycoproteins reverse transcriptase enzyme integrase enzyme protease enzyme Retroviridae The

More information

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors VIROLOGY Engineering Viral Genomes: Retrovirus Vectors Viral vectors Retrovirus replicative cycle Most mammalian retroviruses use trna PRO, trna Lys3, trna Lys1,2 The partially unfolded trna is annealed

More information

Viral Genetics. BIT 220 Chapter 16

Viral Genetics. BIT 220 Chapter 16 Viral Genetics BIT 220 Chapter 16 Details of the Virus Classified According to a. DNA or RNA b. Enveloped or Non-Enveloped c. Single-stranded or double-stranded Viruses contain only a few genes Reverse

More information

Citation for published version (APA): Von Eije, K. J. (2009). RNAi based gene therapy for HIV-1, from bench to bedside

Citation for published version (APA): Von Eije, K. J. (2009). RNAi based gene therapy for HIV-1, from bench to bedside UvA-DARE (Digital Academic Repository) RNAi based gene therapy for HIV-1, from bench to bedside Von Eije, K.J. Link to publication Citation for published version (APA): Von Eije, K. J. (2009). RNAi based

More information

HIV-1 Dual Infection and Neurocognitive Impairment

HIV-1 Dual Infection and Neurocognitive Impairment HIV-1 Dual Infection and Neurocognitive Impairment Gabriel Wagner, MD Assistant Professor of Medicine Infectious Diseases & Global Public Health UC San Diego HIV-Associated End Organ Damage Antiretroviral

More information

Evolution of influenza

Evolution of influenza Evolution of influenza Today: 1. Global health impact of flu - why should we care? 2. - what are the components of the virus and how do they change? 3. Where does influenza come from? - are there animal

More information

Molecular Epidemiology of Simian T-Cell Lymphotropic Virus Type 1 in Wild and Captive Sooty Mangabeys

Molecular Epidemiology of Simian T-Cell Lymphotropic Virus Type 1 in Wild and Captive Sooty Mangabeys JOURNAL OF VIROLOGY, Feb. 2005, p. 2541 2548 Vol. 79, No. 4 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.4.2541 2548.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. Molecular Epidemiology

More information

Far from the limelight of the human and mouse genome projects, the Feline

Far from the limelight of the human and mouse genome projects, the Feline Carolyn Sangokoya Professor Doug Brutlag Genomics and Bioinformatics 9 March 2000 Lessons from the Felidae: A Quest to Understand HIV and other Pathogenic Immunodeficiency Viruses Far from the limelight

More information

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved.

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Apr. 2001, p. 3753 3765 Vol. 75, No. 8 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.8.3753 3765.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Route of Simian

More information

Simian Immunodeficiency Virus SIVagm.sab Infection of Caribbean African Green Monkeys: a New Model for the Study of SIV Pathogenesis in Natural Hosts

Simian Immunodeficiency Virus SIVagm.sab Infection of Caribbean African Green Monkeys: a New Model for the Study of SIV Pathogenesis in Natural Hosts JOURNAL OF VIROLOGY, May 2006, p. 4858 4867 Vol. 80, No. 10 0022-538X/06/$08.00 0 doi:10.1128/jvi.80.10.4858 4867.2006 Copyright 2006, American Society for Microbiology. All Rights Reserved. Simian Immunodeficiency

More information

DATA SHEET. Provided: 500 µl of 5.6 mm Tris HCl, 4.4 mm Tris base, 0.05% sodium azide 0.1 mm EDTA, 5 mg/liter calf thymus DNA.

DATA SHEET. Provided: 500 µl of 5.6 mm Tris HCl, 4.4 mm Tris base, 0.05% sodium azide 0.1 mm EDTA, 5 mg/liter calf thymus DNA. Viral Load DNA >> Standard PCR standard 0 Copies Catalog Number: 1122 Lot Number: 150298 Release Category: A Provided: 500 µl of 5.6 mm Tris HCl, 4.4 mm Tris base, 0.05% sodium azide 0.1 mm EDTA, 5 mg/liter

More information

FENG LI, 1 CAROLINE LEROUX, 1 JODI K. CRAIGO, 1 SHEILA J. COOK, 2 CHARLES J. ISSEL, 2

FENG LI, 1 CAROLINE LEROUX, 1 JODI K. CRAIGO, 1 SHEILA J. COOK, 2 CHARLES J. ISSEL, 2 JOURNAL OF VIROLOGY, Jan. 2000, p. 573 579 Vol. 74, No. 1 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. The S2 Gene of Equine Infectious Anemia Virus Is

More information

The Discovery of SIV and Development of Monkey Models for the Study of HIV/AIDS. Ronald C Desrosiers University of Miami Miller School of Medicine

The Discovery of SIV and Development of Monkey Models for the Study of HIV/AIDS. Ronald C Desrosiers University of Miami Miller School of Medicine The Discovery of SIV and Development of Monkey Models for the Study of HIV/AIDS Ronald C Desrosiers University of Miami Miller School of Medicine 10 FEBRUARY 1984 Infectious Diseases Branch, National

More information

A Comprehensive Panel of Near-Full-Length Clones and Reference Sequences for Non-Subtype B Isolates of Human Immunodeficiency Virus Type 1

A Comprehensive Panel of Near-Full-Length Clones and Reference Sequences for Non-Subtype B Isolates of Human Immunodeficiency Virus Type 1 JOURNAL OF VIROLOGY, July 1998, p. 5680 5698 Vol. 72, No. 7 0022-538X/98/$04.00 0 Copyright 1998, American Society for Microbiology. All Rights Reserved. A Comprehensive Panel of Near-Full-Length Clones

More information

AIDS 111. The origins and diversification of HIV INTRODUCTION THE DEEP ROOTS OF HIV

AIDS 111. The origins and diversification of HIV INTRODUCTION THE DEEP ROOTS OF HIV AIDS 111 The origins and diversification of HIV INTRODUCTION Human immunodeficiency virus (HIV) is not one but several related viruses that have crossed into the human population on multiple occasions

More information

Going Wild: Lessons from Naturally Occurring T-Lymphotropic Lentiviruses

Going Wild: Lessons from Naturally Occurring T-Lymphotropic Lentiviruses CLINICAL MICROBIOLOGY REVIEWS, Oct. 2006, p. 728 762 Vol. 19, No. 4 0893-8512/06/$08.00 0 doi:10.1128/cmr.00009-06 Copyright 2006, American Society for Microbiology. All Rights Reserved. Going Wild: Lessons

More information

Display Unusual Genetic Diversity

Display Unusual Genetic Diversity JOURNAL OF VIROLOGY, Mar. 1990, p. 1086-1092 0022-538X/90/031086-07$02.00/0 Copyright 1990, American Society for Microbiology Vol. 64, No. 3 Simian Immunodeficiency Viruses from African Green Monkeys Display

More information

Under the Radar Screen: How Bugs Trick Our Immune Defenses

Under the Radar Screen: How Bugs Trick Our Immune Defenses Under the Radar Screen: How Bugs Trick Our Immune Defenses Session 7: Cytokines Marie-Eve Paquet and Gijsbert Grotenbreg Whitehead Institute for Biomedical Research HHV-8 Discovered in the 1980 s at the

More information

Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T

Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T cells, the RNA genomes with all modifications are generated

More information

Lecture 12. Immunology and disease: parasite antigenic diversity. and. Phylogenetic trees

Lecture 12. Immunology and disease: parasite antigenic diversity. and. Phylogenetic trees Lecture 12 Immunology and disease: parasite antigenic diversity and Phylogenetic trees Benefits of antigenic variation 2. Infect hosts with prior exposure Hosts often maintain memory against prior infections,

More information

Acquired immune deficiency syndrome.

Acquired immune deficiency syndrome. Acquired immune deficiency syndrome 491 Acquired immune deficiency syndrome. The first cases of acquired immune deficiency syndrome (AIDS) were reported in 1981 but it is now clear that cases of the disease

More information

Molecular Biology and Pathogenesis of Animal Lentivirus Infections

Molecular Biology and Pathogenesis of Animal Lentivirus Infections CLINICAL MICROBIOLOGY REVIEWS, Jan. 1996, p. 100 117 Vol. 9, No. 1 0893-8512/96/$04.00 0 Copyright 1996, American Society for Microbiology Molecular Biology and Pathogenesis of Animal Lentivirus Infections

More information

MINIREVIEW. Simian Immunodeficiency Virus Infection of Chimpanzees

MINIREVIEW. Simian Immunodeficiency Virus Infection of Chimpanzees JOURNAL OF VIROLOGY, Apr. 2005, p. 3891 3902 Vol. 79, No. 7 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.7.3891 3902.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. MINIREVIEW

More information

Mayo Clinic HIV ecurriculum Series Essentials of HIV Medicine Module 2 HIV Virology

Mayo Clinic HIV ecurriculum Series Essentials of HIV Medicine Module 2 HIV Virology Mayo Clinic HIV ecurriculum Series Essentials of HIV Medicine Module 2 HIV Virology Eric M. Poeschla, MD Professor of Medicine College of Medicine Consultant, Division of Infectious Diseases Mayo Clinic

More information

HIV-1 Subtypes: An Overview. Anna Maria Geretti Royal Free Hospital

HIV-1 Subtypes: An Overview. Anna Maria Geretti Royal Free Hospital HIV-1 Subtypes: An Overview Anna Maria Geretti Royal Free Hospital Group M Subtypes A (1, 2, 3) B C D F (1, 2) G H J K Mechanisms of HIV-1 genetic diversification Point mutations RT error rate: ~1 per

More information

Infection of a Yellow Baboon with Simian Immunodeficiency Virus from African Green Monkeys: Evidence for Cross-Species Transmission in the Wild

Infection of a Yellow Baboon with Simian Immunodeficiency Virus from African Green Monkeys: Evidence for Cross-Species Transmission in the Wild JOURNAL OF VIROLOGY, Dec. 1994, p. 8454-8460 0022-538X/94/$04.00+0 Copyright C 1994, American Society for Microbiology Vol. 68, No. 12 Infection of a Yellow Baboon with Simian Immunodeficiency Virus from

More information

HIV Life Cycle & Genetics

HIV Life Cycle & Genetics HIV Life Cycle & enetics! etroviruses (and transposable elements) appear to be part of every cell's genome! From bacteria to yeast, flies, fish, and humans! ome endogenous retroviruses (most notably in

More information

HIV & AIDS: Overview

HIV & AIDS: Overview HIV & AIDS: Overview UNIVERSITY OF PAPUA NEW GUINEA SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY & MOLECULAR BIOLOGY PBL SEMINAR VJ TEMPLE 1 What

More information

Mother-to-Infant Transmission of Simian Immunodeficiency Virus Is Rare in Sooty Mangabeys and Is Associated with Low Viremia

Mother-to-Infant Transmission of Simian Immunodeficiency Virus Is Rare in Sooty Mangabeys and Is Associated with Low Viremia JOURNAL OF VIROLOGY, June 2011, p. 5757 5763 Vol. 85, No. 12 0022-538X/11/$12.00 doi:10.1128/jvi.02690-10 Copyright 2011, American Society for Microbiology. All Rights Reserved. Mother-to-Infant Transmission

More information

Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection

Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection Joseph J. Mattapallil 1, Daniel C. Douek 2, Brenna Hill 2, Yoshiaki Nishimura 3, Malcolm Martin 3 & Mario

More information

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques SUPPORTING INFORMATION FOR: Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques Lia Vassena, 1,2 Huiyi Miao, 1 Raffaello Cimbro,

More information

Feb 11, Gene Therapy. Sam K.P. Kung Immunology Rm 417 Apotex Center

Feb 11, Gene Therapy. Sam K.P. Kung Immunology Rm 417 Apotex Center Gene Therapy Sam K.P. Kung Immunology Rm 417 Apotex Center Objectives: The concept of gene therapy, and an introduction of some of the currently used gene therapy vector Undesirable immune responses to

More information

, virus identified as the causative agent and ELISA test produced which showed the extent of the epidemic

, virus identified as the causative agent and ELISA test produced which showed the extent of the epidemic 1 Two attributes make AIDS unique among infectious diseases: it is uniformly fatal, and most of its devastating symptoms are not due to the causative agent Male to Male sex is the highest risk group in

More information

Host-Specific Modulation of the Selective Constraints Driving Human Immunodeficiency Virus Type 1 env Gene Evolution

Host-Specific Modulation of the Selective Constraints Driving Human Immunodeficiency Virus Type 1 env Gene Evolution JOURNAL OF VIROLOGY, May 1999, p. 3764 3777 Vol. 73, No. 5 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Host-Specific Modulation of the Selective Constraints

More information

A PROJECT ON HIV INTRODUCED BY. Abdul Wahab Ali Gabeen Mahmoud Kamal Singer

A PROJECT ON HIV INTRODUCED BY. Abdul Wahab Ali Gabeen Mahmoud Kamal Singer A PROJECT ON HIV INTRODUCED BY Abdul Wahab Ali Gabeen Mahmoud Kamal Singer Introduction: Three groups of nations have been identified in which the epidemiology of HIV(Human Immunodeficiency Virus) varies:

More information

Determining the Frequency and Mechanisms of HIV-1 and HIV-2 RNA Copackaging by Single-Virion Analysis

Determining the Frequency and Mechanisms of HIV-1 and HIV-2 RNA Copackaging by Single-Virion Analysis JOURNAL OF VIROLOGY, Oct. 2011, p. 10499 10508 Vol. 85, No. 20 0022-538X/11/$12.00 doi:10.1128/jvi.05147-11 Copyright 2011, American Society for Microbiology. All Rights Reserved. Determining the Frequency

More information

Overview: Chapter 19 Viruses: A Borrowed Life

Overview: Chapter 19 Viruses: A Borrowed Life Overview: Chapter 19 Viruses: A Borrowed Life Viruses called bacteriophages can infect and set in motion a genetic takeover of bacteria, such as Escherichia coli Viruses lead a kind of borrowed life between

More information

Human immunodeficiency viruses HIV-1

Human immunodeficiency viruses HIV-1 Origins of HIV and the Evolution of Resistance to AIDS Jonathan L. Heeney, 1 * Angus G. Dalgleish, 2 Robin A. Weiss 3 The cross-species transmission of lentiviruses from African primates to humans has

More information

The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters

The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Emergence of simian immunodeficiency virus in rhesus macaques is characterized by changes in structural and accessory genes that enhance fitness in the new host species The Harvard community has made this

More information

CDC site UNAIDS Aids Knowledge Base http://www.cdc.gov/hiv/dhap.htm http://hivinsite.ucsf.edu/insite.jsp?page=kb National Institute of Allergy and Infectious Diseases http://www.niaid.nih.gov/default.htm

More information

L I F E S C I E N C E S

L I F E S C I E N C E S 1a L I F E S C I E N C E S 5 -UUA AUA UUC GAA AGC UGC AUC GAA AAC UGU GAA UCA-3 5 -TTA ATA TTC GAA AGC TGC ATC GAA AAC TGT GAA TCA-3 3 -AAT TAT AAG CTT TCG ACG TAG CTT TTG ACA CTT AGT-5 OCTOBER 31, 2006

More information

The Evolutionary Rate of Nonpathogenic Simian Immunodeficiency Virus (SIVagm) Is in Agreement with a Rapid and Continuous Replication in Vivo

The Evolutionary Rate of Nonpathogenic Simian Immunodeficiency Virus (SIVagm) Is in Agreement with a Rapid and Continuous Replication in Vivo VIROLOGY 223, 89 102 (1996) ARTICLE NO. 0458 The Evolutionary Rate of Nonpathogenic Simian Immunodeficiency Virus (SIVagm) Is in Agreement with a Rapid and Continuous Replication in Vivo MICHAELA C. MÜLLER-TRUTWIN,*

More information

Lecture 2: Virology. I. Background

Lecture 2: Virology. I. Background Lecture 2: Virology I. Background A. Properties 1. Simple biological systems a. Aggregates of nucleic acids and protein 2. Non-living a. Cannot reproduce or carry out metabolic activities outside of a

More information

Nef-Mediated Enhancement of Virion Infectivity and Stimulation of Viral Replication Are Fundamental Properties of Primate Lentiviruses

Nef-Mediated Enhancement of Virion Infectivity and Stimulation of Viral Replication Are Fundamental Properties of Primate Lentiviruses JOURNAL OF VIROLOGY, Dec. 2007, p. 13852 13864 Vol. 81, No. 24 0022-538X/07/$08.00 0 doi:10.1128/jvi.00904-07 Copyright 2007, American Society for Microbiology. All Rights Reserved. Nef-Mediated Enhancement

More information

Irina Maljkovic Berry

Irina Maljkovic Berry Thesis for doctoral degree (Ph.D.) 2008 Thesis for doctoral degree (Ph.D.) 2008 Genetic aspects of HIV-1 evolution and transmission Irina Maljkovic Berry Genetic aspects of HIV-1 evolution and transmission

More information

JOURNAL OF VIROLOGY, Oct. 1999, p Vol. 73, No. 10. Copyright 1999, American Society for Microbiology. All Rights Reserved.

JOURNAL OF VIROLOGY, Oct. 1999, p Vol. 73, No. 10. Copyright 1999, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Oct. 1999, p. 8201 8215 Vol. 73, No. 10 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Role of Immune Responses against the Envelope

More information

Rajesh Kannangai Phone: ; Fax: ; *Corresponding author

Rajesh Kannangai   Phone: ; Fax: ; *Corresponding author Amino acid sequence divergence of Tat protein (exon1) of subtype B and C HIV-1 strains: Does it have implications for vaccine development? Abraham Joseph Kandathil 1, Rajesh Kannangai 1, *, Oriapadickal

More information

Clinical Significance of Human Immunodeficiency Virus Type 1 Replication Fitness

Clinical Significance of Human Immunodeficiency Virus Type 1 Replication Fitness CLINICAL MICROBIOLOGY REVIEWS, Oct. 2007, p. 550 578 Vol. 20, No. 4 0893-8512/07/$08.00 0 doi:10.1128/cmr.00017-07 Copyright 2007, American Society for Microbiology. All Rights Reserved. Clinical Significance

More information

Received 4 December 2001/Accepted 29 April 2002

Received 4 December 2001/Accepted 29 April 2002 JOURNAL OF VIROLOGY, Aug. 2002, p. 8433 8445 Vol. 76, No. 16 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.16.8433 8445.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. The Relationship

More information

Prokaryotic Biology. VIRAL STDs, HIV-1 AND AIDS

Prokaryotic Biology. VIRAL STDs, HIV-1 AND AIDS Prokaryotic Biology VIRAL STDs, HIV-1 AND AIDS Prokaryotic Biology FROM THE CDC VIRAL STDs, HIV-1 AND AIDS VIRAL STDs & CONTACT VIRAL DISEASES A. GENITAL HERPES & COLD SORES 1. HERPES SIMPLEX VIRUS-2 (HHV-2)

More information